Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 1218: 53-106, 2015.
Article in English | MEDLINE | ID: mdl-25319646

ABSTRACT

This chapter describes how to design and conduct experiments to deliver siRNA to adherent cell cultures in vitro by magnetic force-assisted transfection using self-assembled complexes of small interfering RNA (siRNA) and cationic lipids or polymers that are associated with magnetic nanoparticles (MNPs). These magnetic complexes are targeted to the cell surface by the application of a gradient magnetic field. A further development of the magnetic drug-targeting concept is combining it with an ultrasound-triggered delivery using magnetic microbubbles as a carrier for gene or drug delivery. For this purpose, selected MNPs, phospholipids, and siRNAs are assembled in the presence of perfluorocarbon gas into flexible formulations of magnetic lipospheres (microbubbles). Methods are described how to accomplish the synthesis of magnetic nanoparticles for magnetofection and how to test the association of siRNA with the magnetic components of the transfection vector. A simple method is described to evaluate magnetic responsiveness of the magnetic siRNA transfection complexes and estimate the complex loading with magnetic nanoparticles. Procedures are provided for the preparation of magnetic lipoplexes and polyplexes of siRNA as well as magnetic microbubbles for magnetofection and downregulation of the target gene expression analysis with account for the toxicity determined using an MTT-based respiration activity test. A modification of the magnetic transfection triplexes with INF-7, fusogenic peptide, is described resulting in reporter gene silencing improvement in HeLa, Caco-2, and ARPE-19 cells. The methods described can also be useful for screening vector compositions and novel magnetic nanoparticle preparations for optimized siRNA transfection by magnetofection in any cell type.


Subject(s)
Drug Carriers/chemistry , Magnetite Nanoparticles/chemistry , RNA Interference , RNA, Small Interfering/chemistry , Transfection/methods , Caco-2 Cells , Cell Line, Tumor , Cell Respiration , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fluorocarbons/chemistry , Genetic Vectors , HeLa Cells , Humans , Imines/chemistry , Iodine Radioisotopes , Magnetic Fields , Microbubbles , Phospholipids/chemistry , Plasmids/chemistry , Plasmids/metabolism , Polyethylenes/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ultrasonics
2.
Pharm Res ; 32(1): 103-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25033763

ABSTRACT

PURPOSE: To explore the potential of magnetofection in delivering pDNA to primary mouse embryonic fibroblasts (PMEFs) and porcine fetal fibroblasts (PFFs) and investigate an effect of magnetic cell labeling on transfection efficacy. METHODS: The formulation and a dose of the magnetic vector were optimized. The efficacy of the procedure was quantified by vector internalization, transgene expression and cell iron loading upon specific labeling with Ab-conjugated magnetic beads or non-specific labeling with MNPs. RESULTS: Up to sixty percent of PMEF and PFF cells were transfected at low pDNA doses of 4-16 pg pDNA/cell. Specific labeling of the PMEFs with MNPs, resulted in a 3- and 2-fold increase in pDNA internalization upon magnetofection and lipofection, respectively, that yielded a 2-4-fold increase in percent of transgene-expressing cells. Non-specific cell labeling had no effect on the efficacy of the reporter expression, despite the acquisition of similar magnetic moments per cell. In PFFs, specific magnetic labeling of the cell surface receptors inhibited internalization and transfection efficacy. CONCLUSIONS: Magnetic labeling of cell-surface receptors combined with the application of an inhomogenous magnetic field (nanomagnetic activation) can affect the receptor-mediated internalization of delivery vectors and be used to control nucleic acid delivery to cells.


Subject(s)
DNA/administration & dosage , Drug Carriers/administration & dosage , Fibroblasts/metabolism , Magnetic Fields , Magnetite Nanoparticles/administration & dosage , Transfection/methods , Animals , Cells, Cultured , DNA/genetics , Drug Carriers/chemistry , Magnetite Nanoparticles/chemistry , Mice , Microscopy, Electron, Transmission , Plasmids , Primary Cell Culture , Staining and Labeling , Surface Properties , Swine
3.
Pharm Res ; 29(5): 1344-65, 2012 May.
Article in English | MEDLINE | ID: mdl-22222384

ABSTRACT

PURPOSE: To optimize silica-iron oxide magnetic nanoparticles with surface phosphonate groups decorated with 25-kD branched polyethylenimine (PEI) for gene delivery. METHODS: Surface composition, charge, colloidal stabilities, associations with adenovirus, magneto-tranduction efficiencies, cell internalizations, in vitro toxicities and MRI relaxivities were tested for the particles decorated with varying amounts of PEI. RESULTS: Moderate PEI-decoration of MNPs results in charge reversal and destabilization. Analysis of space and time resolved concentration changes during centrifugation clearly revealed that at >5% PEI loading flocculation gradually decreases and sufficient stabilization is achieved at >10%. The association with adenovirus occurred efficiently at levels over 5% PEI, resulting in the complexes stable in 50% FCS at a PEI-to-iron w/w ratio of ≥7%; the maximum magneto-transduction efficiency was achieved at 9-12% PEI. Primary silica iron oxide nanoparticles and those with 11.5% PEI demonstrated excellent r(2)* relaxivity values (>600 s(-1)(mM Fe)(-1)) for the free and cell-internalized particles. CONCLUSIONS: Surface decoration of the silica-iron oxide nanoparticles with a PEI-to-iron w/w ratio of 10-12% yields stable aqueous suspensions, allows for efficient viral gene delivery and labeled cell detection by MRI.


Subject(s)
Ferric Compounds/chemistry , Gene Transfer Techniques , Genetic Vectors/chemistry , Magnetics , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Adenoviridae/genetics , Animals , Cell Line , Colloids/chemistry , Drug Stability , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Magnetic Resonance Imaging , Mice , Microscopy, Electron, Transmission , Models, Molecular , Polyethyleneimine/chemistry , Rats , Surface Properties , X-Ray Diffraction
4.
Blood ; 117(16): e171-81, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21357765

ABSTRACT

Research applications and cell therapies involving genetically modified cells require reliable, standardized, and cost-effective methods for cell manipulation. We report a novel nanomagnetic method for integrated cell separation and gene delivery. Gene vectors associated with magnetic nanoparticles are used to transfect/transduce target cells while being passaged and separated through a high gradient magnetic field cell separation column. The integrated method yields excellent target cell purity and recovery. Nonviral and lentiviral magselectofection is efficient and highly specific for the target cell population as demonstrated with a K562/Jurkat T-cell mixture. Both mouse and human enriched hematopoietic stem cell pools were effectively transduced by lentiviral magselectofection, which did not affect the hematopoietic progenitor cell number determined by in vitro colony assays. Highly effective reconstitution of T and B lymphocytes was achieved by magselectofected murine wild-type lineage-negative Sca-1(+) cells transplanted into Il2rg(-/-) mice, stably expressing GFP in erythroid, myeloid, T-, and B-cell lineages. Furthermore, nonviral, lentiviral, and adenoviral magselectofection yielded high transfection/transduction efficiency in human umbilical cord mesenchymal stem cells and was fully compatible with their differentiation potential. Upscaling to a clinically approved automated cell separation device was feasible. Hence, once optimized, validated, and approved, the method may greatly facilitate the generation of genetically engineered cells for cell therapies.


Subject(s)
Cell Separation/methods , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Animals , Antigens, Ly/genetics , Genetic Vectors/chemistry , Hematopoietic Stem Cells/metabolism , Humans , Interleukin Receptor Common gamma Subunit/genetics , Jurkat Cells , K562 Cells , Magnetics , Membrane Proteins/genetics , Mesenchymal Stem Cells/metabolism , Mice , Nanoparticles/chemistry , Transfection
5.
Methods Mol Biol ; 605: 487-525, 2010.
Article in English | MEDLINE | ID: mdl-20072903

ABSTRACT

In a magnetofection procedure, self-assembling complexes of enhancers like cationic lipids with plasmid DNA or small interfering RNA (siRNA) are associated with magnetic nanoparticles and are then concentrated at the surface of cultured cells by applying a permanent inhomogeneous magnetic field. This process results in a considerable improvement in transfection efficiency compared to transfection carried out with nonmagnetic gene vectors. This article describes how to synthesize magnetic nanoparticles suitable for nucleic acid delivery by liposomal magnetofection and how to test the plasmid DNA and siRNA association with the magnetic components of the transfection complex. Protocols are provided for preparing magnetic lipoplexes, performing magnetofection in adherent and suspension cells, estimating the association/internalization of vectors with cells, performing reporter gene analysis, and assessing cell viability. The methods described here can be used to screen magnetic nanoparticles and formulations for the delivery of nucleic acids by liposomal magnetofection in any cell type.


Subject(s)
DNA/administration & dosage , Liposomes/chemistry , Magnetics , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , Transfection , Cell Line, Tumor , Cell Survival , Gene Expression , Genes, Reporter , Humans , Iron/chemistry , Plasmids/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...